Abstract

Lysine (K)-specific demethylase 5C (KDM5C) plays a significant role in the tumor cell proliferation, invasion, drug resistance and the regulation of tumor-related gene expression. Here, we aimed to investigate its predictive value in patients with cancers received immune checkpoint inhibitors (ICIs). We explored the predictive value of KDM5C alterations and the association between KDM5C alteration and immune landscape by using published cohort with clinical outcome and sequenced data from online database. The frequency of KDM5C alterations was 2.1% across 48045 tumor samples with different cancers from 185 studies. KDM5C alterations were correlated with markedly inferior overall survival (OS, 53 vs. 102 months, P<0.0001) than those without. However, in ICI-treated group, patients with KDM5C alterations had a substantially prolonged OS than the wild-type group (not reached vs. 18 months, P=0.0041). The predictive value of KDM5C alterations for ICI treatment outcome was not observed in patients with microsatellite-stable tumors (P=0.2875). Intriguingly, patients with non-small-cell lung cancer and KDM5C alterations receiving ICI had the better progression-free survival than wild type group (13.2 vs. 3.2 months, P=0.0762). Mechanistically, KDM5C altered tumors had dramatically higher TMB level and was associated with significantly higher level of CD8+ T cell infiltration and T effector signature. In conclusion, KDM5C alterations was correlated with enhanced tumor immunogenicity and inflamed anti-tumor immunity, thus resulting in better treatment outcome in cancer patients receiving ICIs.

Highlights

  • Immune checkpoint inhibitors (ICIs) targeting cytotoxic T lymphocyte antigen-4 (CTLA-4), or programmed cell death protein 1 (PD-1) and its ligand (PD-L1) interaction have shifted the treatment paradigms and significantly improve the overall survival (OS) in diverse cancers [1,2,3,4]

  • This study firstly reported the frequency of KDM5C alterations and its pan-cancer predictive value to immune checkpoint inhibitors (ICIs) treatment in various cancers

  • We did not observe the association between KDM5C alterations and prolonged OS in patients with MSS solid tumors, suggesting that it may not be suitable for predicting ICI treatment outcome in MSS solid tumors

Read more

Summary

Introduction

Immune checkpoint inhibitors (ICIs) targeting cytotoxic T lymphocyte antigen-4 (CTLA-4), or programmed cell death protein 1 (PD-1) and its ligand (PD-L1) interaction have shifted the treatment paradigms and significantly improve the overall survival (OS) in diverse cancers [1,2,3,4]. Previous studies reported that genetic alterations of KDM5C were common in various types of cancers including breast, colon, ovarian, prostate cancer and so on It plays a significant role in the tumorigenesis, cancer cell proliferation, invasion, drug resistance and the regulation of tumor-related gene expression [12,13,14]. A recent elegant study analyzed the multi-omics data of 823 advanced renal cell carcinoma and found that somatic mutations in KDM5C correlate with high angiogenesis and AMPK/fatty acid oxidation gene expression, which was enriched in ICIs beneficial group. These findings revealed the contribution of KDM5C to antitumor immune response. It is valuable to explore the predictive value of KDM5C alterations for ICIs treatment outcome in multiple cancers

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call